Recombinant Gallid herpesvirus 2 Uncharacterized gene 11 protein (MDV011)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: We prioritize shipping the format currently in stock. However, if you have a specific format requirement, please indicate it in your order notes. We will fulfill your request if possible.
Lead Time
Delivery time may vary depending on the purchasing method and location. For specific delivery timelines, please contact your local distributors.
Note: All protein shipments are made with standard blue ice packs. If you require dry ice shipping, please inform us in advance. Additional fees will apply.
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend centrifuging the vial briefly before opening to ensure the contents are at the bottom. Reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50%, which you can use as a reference.
Shelf Life
Shelf life is influenced by various factors including storage conditions, buffer composition, temperature, and the protein's inherent stability.
Generally, liquid form has a 6-month shelf life at -20°C/-80°C. Lyophilized form has a 12-month shelf life at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is necessary for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type will be determined during the manufacturing process.
The tag type will be determined during production. If you have a specific tag type preference, please inform us, and we will prioritize developing the specified tag.
Synonyms
MDV011; Uncharacterized gene 11 protein
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-85
Protein Length
full length protein
Species
Gallid herpesvirus 2 (strain Chicken/Md5/ATCC VR-987) (GaHV-2) (Marek's disease herpesvirus type 1)
Target Names
MDV011
Target Protein Sequence
MTSERALTLAPGKVSTADIYEADFSFRREFVRQILQREFILFEISMYIIFIVTFCYKIIL FLFRIIPKDLDHRQRNRGRKMSASA
Uniprot No.

Target Background

Subcellular Location
Host membrane; Single-pass membrane protein.

Q&A

What is Gallid herpesvirus 2 and how does it relate to Marek's disease?

Gallid herpesvirus 2 (GaHV-2) is an oncogenic α-herpesvirus that causes Marek's disease (MD), a T cell lymphosarcoma affecting domestic chickens. The virus exhibits a unique oncogenic mechanism where its genome integrates by homologous recombination into the host genome. Through sophisticated modulation of both viral and cellular gene expression, GaHV-2 induces transformation of latently infected cells, making MD a valuable model for studying viral oncogenesis . The virus employs multiple regulatory mechanisms operating at transcriptional, post-transcriptional, and post-translational levels, involving viral and cellular transcription factors, epigenetic modifications, alternative splicing, microRNAs, and post-translational modifications of viral proteins .

How does the Bacterial Artificial Chromosome (BAC) system facilitate MDV research?

The bacterial artificial chromosome (BAC) system has revolutionized research on Marek's Disease Virus by allowing stable maintenance and manipulation of the complete viral genome in Escherichia coli. This methodology involves introducing BAC vector sequences into a specific locus of the MDV-1 genome through homologous recombination . The resulting viral DNA containing the BAC vector can transform E. coli, creating stable colonies harboring the complete MDV-1 genome as F plasmids (MDV-1 BACs) .

The BAC system offers several advantages for MDV research:

  • It enables precise genetic manipulation of the viral genome in bacterial systems

  • It facilitates creation of viral mutants through techniques like one-step mutagenesis using linear DNA fragments amplified by PCR

  • Infectious virus can be recovered by transfecting BAC DNA into chicken embryo fibroblasts

  • It provides a platform for analyzing both essential and non-essential viral genes

This system has been particularly valuable for studying MDV-1, allowing the creation of mutants like the gB-negative virus, representing the first MDV-1 mutant with deletion of an essential gene .

What expression systems are optimal for producing functional recombinant MDV011 protein?

For successful expression of recombinant MDV011 protein, researchers should consider multiple expression systems based on the protein's characteristics. As MDV011 is a small protein (85 amino acids) with potential membrane-associating domains, the following expression strategies are recommended:

Expression SystemAdvantagesConsiderations
E. coli BL21(DE3)High yield, cost-effectiveMay require fusion partners (SUMO, MBP) for solubility
Baculovirus/insect cellsBetter folding of viral proteinsHigher cost, more complex setup
Avian cell linesNative post-translational modificationsLower yields, specialized media requirements

For E. coli-based expression, temperature optimization is critical - lowering induction temperature to 18-20°C often improves solubility. Including fusion tags like His6, GST, or SUMO not only aids purification but can enhance folding and solubility . For membrane-associated proteins, specialized E. coli strains like C41(DE3) or C43(DE3) designed for membrane protein expression may prove beneficial. Purification typically requires a multi-step approach including affinity chromatography, tag removal, and size exclusion chromatography, with careful buffer optimization to maintain protein stability.

How can researchers effectively use BAC technology to study MDV011 function?

Bacterial artificial chromosome (BAC) technology provides a powerful platform for studying MDV011 function through precise genetic manipulation. The following methodology has proven effective:

  • Design a targeting construct containing a kanamycin resistance cassette flanked by homology arms (40-50bp) corresponding to the MDV011 locus

  • Perform two-step Red recombination (en passant mutagenesis):

    • First recombination introduces the selection marker

    • Second recombination removes the marker, leaving no foreign sequences

  • Verify BAC modifications through:

    • Restriction enzyme digestion patterns

    • PCR across the modified region

    • Full-genome sequencing to confirm absence of unwanted mutations

  • Transfect the modified BAC into chicken embryo fibroblasts to reconstitute infectious virus

  • Compare phenotypes between wild-type and MDV011-mutant viruses:

    • Plaque formation and morphology

    • Growth kinetics through multi-step growth curves

    • Cell-to-cell spread capacity

For more sophisticated analysis, researchers can create specific mutations rather than complete deletions, or generate fluorescently tagged versions of MDV011 to track localization during infection. Complementation studies using cell lines expressing wild-type MDV011 can confirm that observed phenotypes are specifically due to MDV011 modification.

What techniques are most effective for analyzing MDV011 expression during viral infection?

To comprehensively analyze MDV011 expression during different phases of viral infection, researchers should employ a multi-technique approach:

For temporal expression analysis:

  • Quantitative RT-PCR with primers specific to MDV011, normalized to both viral (gB) and cellular (GAPDH) controls

  • Samples should be collected at multiple timepoints (2, 24, 48, 72, 96 hours post-infection) to capture immediate-early, early, and late phases

For protein-level detection:

  • Generate specific antibodies against MDV011 using synthetic peptides or recombinant protein

  • Western blotting for protein accumulation during infection

  • Immunoprecipitation coupled with mass spectrometry to identify potential post-translational modifications

For localization studies:

  • Immunofluorescence microscopy with co-staining for cellular compartment markers

  • Super-resolution techniques (STORM or STED) for precise localization

  • Creation of fluorescently-tagged MDV011 using BAC recombineering for live-cell imaging

For in vivo validation:

  • Immunohistochemistry of infected chicken tissues

  • Single-cell approaches like multiplexed immunofluorescence for cell type-specific expression patterns

The combination of these techniques provides comprehensive insights into the expression dynamics and localization patterns of MDV011 throughout the viral life cycle.

How can researchers design experiments to elucidate potential roles of MDV011 in viral oncogenesis?

A comprehensive experimental design to investigate MDV011's potential role in oncogenesis should include:

  • Generate MDV011-deletion mutants using BAC mutagenesis:

    • Complete deletion mutant

    • Point mutations of key residues

    • Domain-specific modifications

  • In vitro transformation studies:

    • Soft agar colony formation assays comparing wild-type and mutant viruses

    • Focus formation in contact-inhibited fibroblasts

    • Analysis of cellular signaling pathways related to transformation (PI3K/Akt, JAK/STAT)

    • RNA-seq to identify differentially expressed host genes in cells infected with wild-type versus MDV011-deficient viruses

  • In vivo pathogenesis studies:

    • Infection of susceptible chicken lines with wild-type and mutant viruses

    • Monitor for tumor development using standardized scoring systems

    • Histopathological examination of affected tissues

    • Quantification of viral loads in blood and feather follicles using qPCR

  • Molecular interaction studies:

    • Co-immunoprecipitation to identify viral and cellular interaction partners

    • ChIP-seq to determine if MDV011 associates with chromatin

    • Proximity labeling (BioID, APEX) to identify the MDV011 interactome in infected cells

  • Comparative analysis with the major viral oncogene Meq:

    • Co-expression studies to evaluate potential cooperation or antagonism

    • Analysis of effects on Meq-regulated cellular genes like Bcl-2

This multi-faceted approach allows researchers to correlate molecular mechanisms with biological outcomes, providing insights into MDV011's potential contributions to the oncogenic process.

What methodological approaches are effective for studying MDV011's potential role in immune evasion?

To investigate MDV011's potential contributions to immune evasion, researchers should implement the following methodological approaches:

  • Develop in vitro immune interaction models:

    • Co-culture systems pairing infected chicken cells with immune cells

    • Flow cytometry to measure MHC-I/II expression, immune checkpoint molecules, and immune activation markers

    • Cytokine profiling using chicken-specific ELISAs or multiplex assays

  • Analyze host-pathogen protein interactions:

    • Immunoprecipitation coupled with mass spectrometry to identify immune-related binding partners

    • Yeast two-hybrid screening against chicken immune signaling components

    • Bimolecular fluorescence complementation to confirm interactions in chicken cells

  • Functional immune evasion assays:

    • Cytotoxicity assays measuring NK cell and CTL killing of infected cells

    • MHC-I antigen presentation efficacy in wild-type versus MDV011-deficient virus

    • Interferon response studies measuring ISG induction

  • In vivo immune response characterization:

    • Comparative analysis of immune cell infiltration in tissues from chickens infected with wild-type versus MDV011-mutant viruses

    • Immunophenotyping of tumor-infiltrating lymphocytes

    • Cytokine profiles in infected birds

Table 1: Key Immune Parameters to Evaluate When Studying MDV011

Immune ParameterTechniqueExpected Result if MDV011 Functions in Immune Evasion
MHC-I expressionFlow cytometryIncreased MHC-I in MDV011-mutant infection
NK cell activationCD107a degranulation assayEnhanced against MDV011-mutant infected cells
Interferon pathwayRT-qPCR for ISGsHigher ISG expression in MDV011-mutant infection
T cell exhaustionPD-1, CTLA-4, LAG-3 expressionReduced in response to MDV011-mutant virus
Viral persistenceViral load quantificationReduced for MDV011-mutant in immunocompetent hosts

These approaches must account for chicken-specific immune features, using appropriate avian-specific reagents rather than mammalian counterparts .

How can comparative genomics inform understanding of MDV011 function across different viral strains?

Comparative genomic analysis of MDV011 across different strains of Marek's disease virus provides valuable insights into its functional significance and evolution. An effective methodological approach includes:

  • Sequence alignment and conservation analysis:

    • Align MDV011 sequences from highly virulent (e.g., RB1B, Md5), mildly virulent (e.g., JM), and vaccine strains (e.g., CVI988/Rispens)

    • Identify conserved domains that likely serve essential functions

    • Map strain-specific variations that may correlate with virulence differences

  • Evolutionary analysis:

    • Calculate selection pressures (dN/dS ratios) across the protein to identify regions under positive or purifying selection

    • Phylogenetic analysis to determine evolutionary relationships between MDV011 variants

    • Identify potential recombination events that may have affected MDV011 evolution

  • Structural prediction and comparison:

    • Generate structural models of MDV011 from different strains using tools like AlphaFold

    • Compare predicted structures to identify conserved structural elements despite sequence variations

  • Functional validation through chimeric viruses:

    • Use BAC technology to create recombinant viruses where MDV011 from virulent strains is replaced with counterparts from vaccine strains

    • Assess phenotypic changes in replication, cell tropism, and pathogenicity

    • Perform transcriptome analysis to identify differential host responses to different MDV011 variants

This comprehensive approach not only characterizes MDV011 function but may identify strain-specific features that could be targeted for improved vaccine development against more virulent emerging strains .

What are the potential approaches for targeting MDV011 in vaccine development?

Developing vaccines targeting MDV011 requires understanding its role in viral pathogenesis and applying this knowledge to vaccination strategies. Potential approaches include:

  • Subunit vaccine development:

    • Express recombinant MDV011 protein with appropriate adjuvants

    • Design peptide vaccines based on MDV011 epitopes predicted to be immunogenic

    • Evaluate antibody production and cellular immune responses

  • Live-attenuated vaccine strategies:

    • Use BAC technology to create MDV011-modified viruses with attenuated pathogenicity but retained immunogenicity

    • Consider conditional expression systems where MDV011 function is temperature-sensitive

    • Evaluate protection efficacy against challenge with virulent field strains

  • DNA vaccine approaches:

    • Develop plasmids encoding MDV011 under strong promoters

    • Optimize codon usage for expression in chicken cells

    • Consider prime-boost strategies combining DNA and protein immunization

  • Vectored vaccine platforms:

    • Express MDV011 in fowlpox or herpesvirus of turkeys (HVT) vectors

    • Evaluate the benefit of co-expressing MDV011 with other immunogenic MDV proteins

    • Assess cellular and humoral immune responses

The efficacy of these approaches depends on determining whether MDV011 represents a protective antigen, which requires comprehensive immunogenicity studies and challenge experiments in the natural host.

How might single-cell approaches advance understanding of MDV011 function in disease pathogenesis?

Single-cell technologies offer unprecedented opportunities to dissect the role of MDV011 in the heterogeneous cellular landscape of Marek's disease pathogenesis:

  • Single-cell RNA sequencing applications:

    • Compare transcriptomes of individual cells infected with wild-type versus MDV011-deficient viruses

    • Identify cell populations differentially affected by MDV011 expression

    • Map infection trajectories from early infection to transformation at single-cell resolution

  • Single-cell proteomics approaches:

    • Mass cytometry (CyTOF) with metal-conjugated antibodies against viral and cellular proteins

    • Imaging mass cytometry of infected tissues to preserve spatial context

    • Proximity labeling at single-cell level to identify MDV011 interaction partners

  • Spatial transcriptomics methods:

    • Visualize MDV011 expression in the context of tissue microenvironment

    • Correlate MDV011 expression with local immune cell infiltration

    • Identify niches where MDV011 may play specific roles in immune evasion or transformation

  • Integrated multi-omic approaches:

    • Combined single-cell RNA-seq and ATAC-seq to correlate MDV011 expression with chromatin accessibility changes

    • Single-cell BCR/TCR sequencing to track clonal expansion of B and T cells in response to MDV011-positive versus MDV011-negative viruses

These advanced approaches can reveal cell type-specific effects of MDV011 that might be masked in bulk analyses, providing insights into how this protein contributes to the complex pathogenesis of Marek's disease.

What structural biology approaches might advance understanding of MDV011 function?

Structural biology offers powerful tools to illuminate the function of uncharacterized proteins like MDV011:

  • Computational structure prediction:

    • Employ machine learning approaches like AlphaFold2 to predict MDV011 structure

    • Identify potential functional domains, secondary structure elements, and surface features

    • Predict protein-protein interaction sites through computational docking

  • X-ray crystallography approach:

    • Express and purify milligram quantities of MDV011 with high purity

    • Screen crystallization conditions using commercial sparse matrix screens

    • Optimize promising conditions to obtain diffraction-quality crystals

    • Determine atomic resolution structure through X-ray diffraction

  • Nuclear Magnetic Resonance (NMR) spectroscopy:

    • Particularly suitable for MDV011 due to its small size (85 amino acids)

    • Allows structure determination in solution

    • Can provide insights into dynamic regions and conformational changes

  • Cryo-electron microscopy:

    • Valuable for visualizing MDV011 in context of larger complexes

    • May reveal structural details of MDV011 interaction with viral or cellular partners

  • Functional validation of structural insights:

    • Site-directed mutagenesis of key residues identified in the structure

    • Biochemical and virological assays to assess impact on protein function and viral fitness

    • Hydrogen-deuterium exchange mass spectrometry to map interaction interfaces

The integration of structural data with evolutionary analysis across different viral strains can identify conserved functional motifs versus strain-specific features, informing both basic understanding of MDV011 function and potential applications in vaccine or antiviral development.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.